Category Archives: #ASCO14

PD-1 Pathway Inhibition: Moving Beyond Melanoma

As we watch the clinical development of PD-1 pathway inhibitors we are struck by the ability of this approach to produce clinical efficacy in diverse cancers. Here we will briefly run through the rest of the landscape, starting with non-small cell lung cancer (NSCLC), then touching on metastatic renal cell carcinoma (RCC), glioblastoma (GBM), bladder cancer (UB), ovarian carcinoma and others. In many cases we are beginning to see the use of combination therapy, a setting that generally requires a careful look at toxicity. We’ll also look at the contentious issue of biomarker development for PD-1 and PD-L1 antagonists. The data are broken down into easy to understand bits, otherwise the whole thing is overwhelming.

The Emerging Role of PD-1 Pathway Inhibition in Lung Cancer

In the context of the often brutally aggressive tumors classified as the Non-Small-Cell Lung Cancers (NSCLC) progress is difficult and even incremental improvement in care is cause for celebration. Even so it was difficult to gather a consensus view of the clinical data being generated by the PD-1 pathway antibodies in NSCLC. In the melanoma setting (parts 1 and 2) our enthusiasm for PD-1 pathway inhibitors is driven by really terrific responses in some patients. The goal there is to improve the response rates while controlling toxicity. The clinicians argue, certainly with merit, that the responses seen justify the occasionally difficult toxicity. After all, all of their available therapies are limited by toxicity concerns, not only chemotherapy but also the small molecule targeted therapeutics. In this context, PD-1 directed therapies are well positioned.

It is not yet clear how these issues will play out in NSCLC. On the positive side, a subset of patients respond remarkably well to PD-1 pathway inhibition, and we may develop an understanding of how identify such patients. However, the overall response rates remain low, and impact on PFS and OS is small (see part 1 for a list of abbreviations). Complicating our understanding of the benefit of this class of therapeutics in NSCLC are three observations. The first observation is that tumor responses in this disease setting can be anomalous and may not be appropriately captured by standard RECIST tumor response criteria. The second observation is that severe toxicity can truly derail patients, and may even sensitize some patients to chronic toxicity that prevents application of other types of therapies. The third observation is that some targeted therapeutics for molecularly defined subsets of NSCLC patients are an attractive option to PD-1 directed therapeutics, and we don’t know yet if these can be combined.

The amount of data presented at ASCO14 was huge, and won’t try to cover it all.

There was a pretty dramatic response to the clinical trial data describing the utility of PD-1 pathway inhibitors in NSCLC. Bristol-Myers Squibb’s (NASDAQ: BMY) stock price dropped more than 6% from the start of ASCO on May 30th through the following week, and this on top of a long slide that started in March. Merck & Co (NYSE: MRK) stock jumped more than 2.5% at the same time. What was going on here? Let’s look at the estimated market sizes for three critical indications:Screen Shot 2014-06-18 at 6.55.32 PM

It is clear from the table that NSCLC is the largest patient population by more than 10-fold. It is also the case that NSCLC lacks the range of treatment options available to advanced melanoma and RCC patients. From the investor perspective then, NSCLC is a very big deal. Lets start there, and see how the emerging PD-1 pathway therapeutic class did in this setting.

Nivolumab anti-PD-1 antibody was studied in a number of clinical settings (ASCO14 abstracts #8024 and #8113). As monotherapy, nivo was generally well tolerated, and very effective for some patients. The ORR = 22% – 36% and a subset of patients had a durable response. Combining nivo with dual-platinum based chemotherapy increased the response rate but dramatically increased the SAEs, and in this early data did not appear to impact 1 year survival rates significantly. This table sums up some of the available data.

Screen Shot 2014-06-18 at 6.57.08 PM

Some patients responded very well – these were typically patients that had a non-squamous cell phenotype and whose tumors expressed PD-L1 and were therefore actively shutting down T cell responses by binding PD-1 on T cells. This figure is from a poster presented by Scott Gettinger (ASCO14 Abstract #8024).

Screen Shot 2014-06-19 at 8.05.11 AM

Tumor size is given on the Y-axis and duration is shown across the X-Axis, so for some responding patients the outcome is very good. In an effort to boost the effectiveness of immunotherapy for NSCLC, BMY ran a combination trial of nivo + ipi, as they had done previously in melanoma. They enrolled chemo-naive NSCLC patients, stratified by cell type into squamous or non-squamous groups, then gave these patients an induction regimen of nivo + ipi for 84 days (4 x 21 day cycles). The doses were either 1 + 3 mpk IV Q3W or 3 + 1 mpk IV Q3W, referring to the dose or nivo + ipi, respectively. Then the patients went onto a nivo maintenance schedule. The combination worked rather well, seeming to wipe out the disparity between the two cell types and overcoming some of the resistance seen in PD-1 tumors. Here are some illustrations of the data from Scott Antonio’s poster (ASCO14 Abstract #8023):

Screen Shot 2014-06-18 at 7.01.29 PM

As you can see, a fair number of patients have a sustained decrease in tumor volume or a stable disease course (no change in tumor volume over time). That’s a very nice result. Now the bad news. This combination therapy was nasty, a toxic brew. Discontinuation rates averaged 35% across the treatment arms with AEs including pneumonitis, liver damage, colitis, autoimmune nephritis (kidney inflammation & fibrosis), pulmonary hemorrhage, endocrinopathy, neuropathy, etc. Six patients (12%) died. We note that only 3 fatalities were directly attributed to the study drugs, but no one felt comfortable with these results. BMY’s stock price promptly dropped. We won’t know if the reaction was justified until other dosing and combo regimens are tried, but investors found another home, and that was with the competing drugs from Merck and Astra Zeneca (NYSE: AZN).

Immediately striking were the results from an NSCLC trial with the  anti-PD-1 antibody  pembrolizumab as monotherapy. Patients were selected based on positive PD-L1 staining on > 1% of tumor cells and given 2 or 10 mpg pembro Q3W or 10 mpk Q2W (ASCO Abstract #8077). The response rate was 26% or more, and the SAE rate was low (4%) although 18% dropped out due to AEs of any grade. Responses were durable with more than half of responders still on treatment at the time of data lock. A couple of things to note: these were treatment-naive patients, so presenting early in disease course. Second this figure (from the presentation by Naiyer Rizvi, ASCO #8007) shows maximum responses (ie. best response) and would not capture rebounding tumor size.

Screen Shot 2014-06-18 at 7.04.35 PM

As side note: the authors introduced an “immune response” criteria response rate because some patients would respond after having a new lesion appear (which would trigger the progressive disease (PD) score). Using these new criteria responses were even higher. We’ll see if these become more widely accepted.

AZN/Medimmune presented early expansion data on their anti-PD-L1 antibody, MEDI4736 (ASCO14 Abstract #3002 presented by Neil Segal) in advanced solid tumors. 84 patients with NSCLC were enrolled. The reported ORR was low but this was an accident of sampling as most patients had not gotten to their second screen yet and could not be scored as responders, per protocol. More impressively the vast majority of patients, across diverse tumor classes, remained on therapy.

Screen Shot 2014-06-18 at 7.06.47 PM

While it will be critical to see this data updated, the early read is very encouraging. Not waiting, AZN has initiated a pivotal trial in NSCLC and also a combo trial with their own anti-CTLA4 antibody, tremelimumab. We should be cautiously optimistic that the combination of anti-PD-L1 antibody with anti-CTLA4 antibody will have fewer tox issues than the anti-PD-1 combination, as mild tox appears to be a common feature of targeting the ligand rather than the receptor.

At this point of development, nivo has run into some problems in the combination setting, pembro looks promising, and MEDI4736 also looks promising. It will be very interesting to follow these story lines as they mature.

OK, RCC and other tumor types next, stay tuned.

PD-1 Pathway Inhibitors & Cancer Therapy – PART 2

Other PD-1 pathway therapeutics in advanced melanoma therapy.

Yesterday we focused on nivolumab, particularly in combination with ipilimumab, for the treatment of advanced melanoma. There are competing PD-1 pathway inhibitors that have now reported out substantial trial data. See part 1 for a list of PD-1 pathway therapeutics in development. Much attention has gone to Merck’s pembrolizumab, formally called MK-3475. The activity of pembrolizumab in melanoma is very similar to that of nivolumab, so it’s worth taking a closer look at the characteristics of the antibodies. Included here is pidilizumab, another anti-PD-1 antibody, developed by CureTech.

Screen Shot 2014-06-13 at 3.29.36 PM

Attributes of note include the different sources of the antibodies (fully human vs humanized murine antibody), different isotypes (IgG4 vs IgG1) and affinities ranging more than 200-fold from sub-100pM to 20nM. However, this is a small number of antibodies and it will be hard to discern how each of these attributes contributes to efficacy. Pembrolizumab closely resembles nivolumab except that the affinity for PD-1 is as much as 10 fold better. At the doses given it is difficult to know if this makes any difference, as drug levels may be saturating. We’d have to dig out target occupancy data from the trials to figure this out, but let’s look at the pembrolizumab results first, as it will become clear that this antibody has similar efficacy as nivolumab. How these therapeutics are being developed is different, as we’ll see.

The pembrolizumab (“pembro”) data reported at ASCO are from a huge Phase 1 clinical trial in advanced melanoma. Importantly, Merck made the strategic decision to stratify patients by prior exposure to the anti-CTLA4 antibody ipilimumab (“ipi”), from Bristol-Myers Squibb. This gave the company a jump on the field, allowing them to pursue FDA approval first for ipi-refractory patients. Due in part to the toxicity associated with ipi therapy, there are a lot of these patients. First, though, a brief look at the data, which has been widely reported. The data are compared to published data for nivolumab (“nivo”) treatment of ipi-naive advanced melanoma patients                 (http://jco.ascopubs.org/content/32/10/1020.long). A guide to the clinical abbreviations is included in part 1.

Screen Shot 2014-06-13 at 3.29.46 PM

If we focus on the ipi-naive ORR and 1 year survival data I think we have to conclude that these drugs are pretty comparable, and we’ll wait for additional data before trying too hard to differentiate these. That data will have to come from longer duration of ongoing trials and various combination studies. It is clear from the monotherapy data is that for advanced melanoma patients, anti-PD-1 therapeutics offer a chance at extended benefit. If we look more closely however,we see that in the nivo trial referenced above, half of the responding patients stopped therapy for reasons other than disease progression, most likely dropping off study due to AEs. It is true that 3/4s of the nivo patients stopping therapy maintained a response, some for extended periods. In the pembro study, the SAE rate was 12% but only 4% of patients discontinued therapy as a result of AEs, so that’s good. The catch is that in order to move ORR higher than 40%, combination therapy may be needed. As we saw with the ipi/nivo combo, this comes with much higher toxicity and drop-out rates. Of course the hope is that moving to earlier line therapy will boost response rates with the same or less toxicity and that data will come with time. As an aside, the question of ORR is the reason we have basically ignored the anti-PD-1 antibody pidilizumab, which had a 5-6% ORR. The 1 year OS was similar to the other anti-PD-1 therapeutics, but with such a low ORR it’s hard to believe this therapeutic from Curetech will gain much traction.

Anti-PD-L1 antibodies constitute the second class of therapeutics targeting the PD-1 pathway. These are in early clinical development in multiple tumor types, and will be addressed later. PD-L1 is also important in the context of predicting response to therapy in melanoma, and the utility of this marker as well as PD-1 is the subject of considerable discussion. When the ORR is 40%, it is helpful to select patients prospectively. We can take a close look at one of the smaller cohort studies to get a good look at this. In a study of responsiveness to pembro, Richard Kefford et al (abstract #3005) used an analysis of PD-L1 expression to demonstrate a remarkable difference in clinical response between patients who had > 1% tumor PD-L1 expression versus those who were PD-L1 negative. Biopsy was required in the 2 months preceding the start of pembro therapy; tumor PD-L1 expression was assessed by immunohistochemical staining. Patients received pembro at either 10 mg/kg Q2W, 10 mg/kg Q3W or 2 mg/kg Q3W. With a median treatment time of 23 weeks and ≥13 months follow-up, ORR was 41%, median PFS was 31 weeks and median OS was not reached. The 1-year survival rate was 81%, so this was a terrific cohort within the larger pembro study, likely due to the higher doses used. PD-L1 expression was associated with improved ORR by (51% vs 6%), PFS (median 12 vs 3 months) and 1-year survival rate (84% vs 69%). Note that while there were no treatment-related deaths; 14% of patients experienced drug-related SAEs (grade 3/4) again reflecting the aggressive dosing schedule.

In the large trial of ipi-naive patients treated with nivo, PD-L1 positive tumor staining was associated with ORR, but only weakly with PFS and OS. Why the data are less robust than the Kefford study is unclear. What is abundantly clear however is that there were profound responses in patients scored as PD-L1 negative, as shown in this screen grab from Dr Weber’s Discussant review of the melanoma oral poster session:

Screen Shot 2014-06-13 at 4.09.22 PM

These data suggest that caution should be exercised in the use of PD-L1 staining as a prognostic tool, and the search for better biomarkers of response continues.

We will revisit some of these issues as we move on to NSCLC, RCC, bladder, ovarian and solid tumors more generally.

PD-1 Pathway Inhibitors Reveal Unique Benefit/Risk Profiles Across Cancer Indications

Introduction

Anyone attending the immunotherapy sessions at ASCO earlier this month would have heard several distinct messages about PD-1 pathway inhibition in oncology. PD-1 appears to be a central control point for curtailing T cell responses in the peripheral tissues, similar to the role that CTLA4 plays in regulating initial T cell activation in secondary lymphoid organs such as the lymph nodes and spleen. Remarkable progress has been made in the 13 years since Gordon Freemen and colleagues first proposed in Nature Immunology that the PD-1 pathway was used by tumor cells as a shield against immune system attack (http://www.ncbi.nlm.nih.gov/pubmed/11224527).

It is clear that PD-1 pathway antagonists show tremendous promise in treating diverse cancers. Less clear is an understanding of why certain patients respond or don’t, what biomarkers might predict response, how to increase response rates, how to accurately measure response, and how to safely combine PD-1 pathway inhibition with other therapies.

Table 1 lists the PD-1 therapeutics in development (some of these therapeutics did not have updates at ASCO).

 Screen Shot 2014-06-12 at 3.52.08 PM

As the table demonstrates, the PD-1 pathway inhibitors are being developed in diverse tumor types. As late Phase 2 data and Phase 3 data are coming out we can begin to see the real promise of these drugs in clinical responses measured in large numbers of patients. The amount of data presented at ASCO was a bit overwhelming so to simplify the landscape we can address each tumor type individually, when possible. Some terms we will use are given in the table below.

Table 2 defines the RECIST1.1 clinical response parameters and their abbreviations.

Screen Shot 2014-06-12 at 3.54.35 PM

To put these terms in perspective we can just consider that a meaningful clinical response is a measureable response to therapy (SD < PR < CR) that is durable and leads to an increase in PFS, which in turn allows a significant increase in OS. There are other terms used to describe clinical responses but these are the most common. We will start with some of the most recent data, and see where that takes us.

Part 1: Immune Checkpoint Combination Treatment of Melanoma 

The very first trials of PD-1 pathway inhibitors began with the investigation of nivolumab in metastatic melanoma. As such, there was an impressive amount of progress reported and we now have mature data on different therapeutics. To set the stage, we can consider the benefit shown by nivolumab monotherapy compared to standard of care treatment protocols, and also to ipilimumab (brand name Vervoy) an anti-CTLA4 antibody, also from Bristol-Myers Squibb (BMY). Ipilimumab is approved for the treatment of metastatic melanoma based on Phase 3 clinical trial data in metastatic melanoma patients that had failed prior therapy (a chemotherapy regimen). The trial compared ipilimumab to a tumor vaccine targeting the melanoma antigen gp100. Ipilimumab treatment improved median OS to 10 months versus 6 months with the vaccine treatment (which was no better than standard of care). The 1 year survival rate was 45%. ORR however was low, just about 10%. Also, adverse events (AEs) were a problem, and included autoimmune manifestations (colitis, pituitary inflammation) and some treatment-related deaths (2% of patients). In a separate study of treatment-naive metastatic melanoma patients, ipilimumab therapy was associated with an OS = 11.2 months and a 1 year survival rate of 47%, falling to 21% by year 3. Patients were given ipilimumab or placebo plus chemotherapy (dacarbazine), and then moved to ipilimumab or placebo alone if there was a response measured or if the initial therapy caused toxicity. One consequence of this scheme was that AEs went up dramatically, with 38% of patients experiencing an immune related, grade 3 or 4 severe AE (SAE). We dwell on the anti-CTLA4 antibody ipilimumab because it is the benchmark for other immunotherapies such as nivolumab.

Nivolumab therapy for advanced melanoma has produced impressive data, with median OS = nearly 17 months, and 1 and 2-year survival rates of 62% and 43%. ORR was 33%. AEs were significant if less severe than those seen with ipilimumab. Grade 3-4 treatment-related AEs were seen in 22% of nivolumab-treated patients. Immune-related adverse events (all grades) were seen in 54% of treated patients, and included skin, GI and endocrine disorders. However only 5% of patients experienced immune-related SAEs of grade 3 or 4 and there were no drug-related deaths. These data from Topalian, Sznol et al. from John Hopkins University School of Medicine were presented at ASCO last year and published earlier this year                       (http://jco.ascopubs.org/content/early/2014/03/03/JCO.2013.53.0105.full.pdf).

So with that as our backdrop lets update the state of PD-1 pathway antagonism in melanoma. One of the obvious next steps in the development of immunotherapy is to combine treatments and we saw dramatic long-term data from the combination trial of ipilimumab plus nivolumab in advanced melanoma. Early trial results presented at ASCO last year introduced 4 cohorts of patients given different doses of nivolumab and ipilimumab in combination, with an ORR across all four cohorts of 40% and a 1 year survival rate of 82%. Median OS had not been reached. SAE rate across the 4 cohorts was 53%. This quickly gets complicated so let’s define the cohorts. Numbers are doses of nivolumab and ipilimumab, respectively, in mg/kg: Cohort 1 (0.3 + 3), Cohort 2 (1 + 3), Cohort 3 (3 + 1), Cohort 4 (3 + 3). No data were presented for Cohorts 6 and 7 so we’ll skip those. Cohort 8 is designed to mimic the dose schedule chosen for later clinical trials.

Note that after the induction phase, patients are moved onto maintenance therapy, as show below.

Screen Shot 2014-06-12 at 3.56.50 PM

The slide is taken from the trial update presented at ASCO by Dr Sznol (Abstract #LBA9003). The data updates drove home several critical points. First, at the optimal dose rates of 1 + 3 and 3 + 1 the ORR ranged from 43-53%. The author’s introduce a new classification of clinical response to capture the observation that many patients are experiencing benefit while not strictly meeting RECIST1.1 criteria, this is termed “Aggregate Clinical Activity Rate” and reaches 81-83% in Cohorts 3 and 4 (note that Cohort 4 (3 + 3) was the maximum tolerated dose due to SAEs and will no longer be used). Perhaps more meaningfully, the percent of patients whose tumor burden was reduced by > 80% at 36 weeks was 42% across the cohorts. This is a remarkable number suggesting sustained clinical benefit. Indeed, in those patients who responded, the median DOR in Cohorts 1-3 plus Cohort 8 has not been reached. In Cohorts 1-3, 18/22 patients are still responding and 7 of those had discontinued therapy due to AEs (more on this below).

Dose cohorts were analyzed for impact on 1 and 2 year survival. In Cohorts 2-3 the 1 year OS = 94% and the 2 year OS = 88%. Most stunning of all was this data showing a median OS in Cohorts 1-3 of 40 months. Median OS in Cohort 3 (1 + 3) has not yet been reached.

Screen Shot 2014-06-12 at 3.58.38 PM

These data are best-in-class for treating advanced melanoma, and place ipilimumab plus nivolumab at the forefront of therapeutic options for these patients. The one outstanding issue remains that of toxicity. 23% of patients had to discontinue therapy due to toxicity, and one patient died of complications resulting from treatment. While Dr Sznol repeatedly pointed out that the toxicities observed are controlled by standard interventions, the problem is that these standard interventions include cessation of therapy. We have already learned from the ipilizumab experience that responses to immune checkpoint inhibition can take time, and for those patients who have to stop treatment after 1 – 2 doses due to toxicity, time may not be kind. It will certainly be beneficial to reduce SAEs so that more patients can remain on therapy.

Tomorrow we’ll look at other PD-1 pathway therapeutics and combinations in melanoma before moving on to other tumor types.

SugarCone Biotech LLC will be at ASCO 2014

SugarCone Biotech LLC will be represented at ASCO by Paul D Rennert, Founder & Principal of the Company. SugarCone is actively sourcing innovative therapeutic programs that address unmet needs in cancer treatment. Our goal at ASCO is to continue to build upon our deep expertise in understanding the drug development landscape across tumor classes. While we focus intently on biologics and immune checkpoint therapeutics, we also have intensive experience with small molecule drug development programs. This unique breadth of clinical expertise, deep tumor biology experience and comprehensive understanding of different therapeutic classes has placed us in the center of the recent surge in successful cancer drug development. Specifically, SugarCone brings programs to partners who are committed to the clinical development of important new therapeutics. We also specialize in working closely with preclinical companies to nurture and mature their drug development programs in order to effectively address the evolving clinical landscape.

We have been very pleased to see our clients successfully develop partnerships with top tier biopharma, including Celgene, Bristol-Myers Squibb and Immunogen. Such success provides well-deserved validation, and the capital to build for the future.

Contact us at info@sugarconebiotech.com or cell/text us at 1-508-282-6370.

See you in Chicago!

 

Targeting TNFRs with agonist antibodies for cancer therapy: 4-1BB, GITR, CD27

One of the puzzles in thinking about the available costimulatory receptors on T cells (and NK cells) is the unsettling number of them. Sticking just to the TNF receptor superfamily (TNFRSF) we have OX40 (discussed earlier), 4-1BB, GITR, CD27, and also the TNF receptors themselves (1 and 2), the lymphotoxin beta-receptor, HVEM, and TNFRSF25. There may be some I’ve forgotten. As noted in part 1 OX40, GITR, 4-1BB and CD27 are evolutionary cousins, as are their cognate ligands. Why did the immune system evolve such a complexity of T cell costimulators?

The answer is not entirely clear although the expression patterns and kinetics of expression suggest some rationale for understanding the number of different receptors. Also, as it’s understood that all the TNF receptors signal via NF-kB, Jun and p38, we might see these receptors either compete (for signaling proteins) or cooperate. All of the available genetic and pharmacologic data suggest they cooperate or even synergize, thereby powering the T cell response when needed. Since T cell responses (and immune responses generally) are so dangerous when dysregulated, the multiplicity of on and off switches presumably allows for redundancy of control.

As we said previously, OX40 comes on slow and easy, starting about 12 hours after TCR stimulation, and riding along for up to 96 hours. This is in vitro, cell culture data … so lets recognize that in vivo, in response to the chaotic presentation of antigen, the population of T cells is likely to be turning over, proliferating … so it’s unlikely we will see a finely tuned kinetic response in the real world, as regards the population of responding cells. Nonetheless we can focus on a single T cell, just the one. And we’ve guessed it will be expressing OX40 say from day 2 to day 5 after activation. Lets ignore the fact that activated effector T cells are likely dividing more rapidly than every 5 days, and just ask the simple question – of the other TNFSF receptors, what else is expressed and when, and on what T cell and other cell types?

Cue 4-1BB.

4-1BB is also expressed following T cell activation, and is expressed on other cell types also. 4-1BB expression come up much more quickly after T cell activation, within a few hours, and wanes after several days. This receptor is critical to supporting activated T cell proliferation, differentiation and survival. Expression on T cells may be coincident with OX40, but the consequences of engaging the receptor with an agonist antibody are different. 4-1BB preferentially supports the proliferation and survival of CD8+ T cells, although at least in some settings the activity of CD4+ T cells is also stimulated through 4-1BB. Much of the anti-tumor activity of 4-1BB agonist antibodies in preclinical studies can be traced to stimulation of NK cells although this depends of the tumor type and model used. Less well understood is the role on 4-1BB on other cells types. This receptor is also found on DCs, macrophages, granulocytes, and Tregs. Expression has also been described on vascular endothelium and on some tumor cells. The role of 4-1BB is confusing, with various studies showing expansion of the Treg subset and others suggesting that 4-1BB dampens Treg responses, perhaps via direct effects on DCs. The 4-1BB gene-knockout mouse shows aspects of autoimmune disorders (at least in the mouse strains tested), suggesting a role for 4-1BB in maintaining immune homeostasis following activation. 4-1BB knockout mice have trouble handling tumor challenge, and at least some spontaneously develop B cell lymphomas as they age. It is all a bit complicated.

Regardless, there are two antibodies that can provide some early clinical data. Bristol-Myers Squibb (BMY) started development of the BMS-663513 antibody quite early, before the immunotherapy wave had really gotten started. BMS-663513 is a specific anti-4-1BB agonist antibody, isotype IgG4. The antibody ran into toxicity issues in a phase 2 trial of metastatic melanoma in 2011, leading to a halt of three trials with that antibody. As the toxicity was correlated with dose, BMY has restarted the clinical campaign with BMS-663513, now called urelumab to establish a safe and efficacious dose. A monotherapy trial is being run in patients with advanced/metastatic solid tumors or with relapsed/refractory Non-Hodgkin Lymphoma (NHL). A second trial in NHL is being run in combination with rituximab treatment. A third trial in advanced/metastatic colorectal and head and neck cancers is being run in combination with cetuximab (anti-EGFR). Pfizer’s (PFE) PF-05082566 is an agonist anti-4-1BB antibody (IgG2 isotype). One trial is listed at clinicaltrials.gov, a 3×3 dose escalation phase 1 trial run as monotherapy in patients with advanced cancers, and as combination therapy with rituximab in NHL.

At the June ASCO meeting there will be updates on both the BMY and PFE programs. BMY has a presentation focused on mechanism of action and biomarker analyses. Abstract #3017 outlines the goal of monitoring the immune status of 4 patients prior to and during the phase 1 study of urelumab (BMS-663513: clinical trial NCT01471210). The antibody was given every 3 weeks and the analysis presents results through 3 cycles. PBMCs were isolated from whole blood, and stimulated for 4 hours with PMA/ionomycin to activate lymphocytes. There was an increase in CD8 T cells up to 41% and NK cells up to 62%. CD4 T cells decreased by as much as 23% and regulatory CD4 T cells decreased by as much as 18% comparing the 3rd cycle to baseline. The results are consistent with a preferential impact on CD8+ T cells and NK cells. The level of the cytokines GM-CSF and IFNgamma were increased.

The PFE study (abstract #3007) describes very early data from clinical trial NCT01307267. Patients received PF-05082566 IV every 4 weeks (one cycle) with an 8 week period for assessment of dose-limiting toxicity (DLT) and radiographic analysis of tumor burden (RECIST 1.1). 27 patients were up to 0.3 mg/kg, the highest dose reported in the abstract. The majority of patients had either colorectal cancer (n=11) or Merkel cell carcinoma (n=6), the rest were a collection of solid tumor patients and 2 patients with B cell lymphoma.  25/27 patients completed the DLT assessment period (first cycle). No DLT was established but only 7 patients remain on therapy. All discontinuations from treatment were due to disease progression. A best overall response of stable disease was observed in 6 patients. No duration data is supplied.

These two early trials suggest that safe dose levels can be achieved, that a mechanism of action can be confirmed (urelumab: expansion of CD8+ T cells and NK cells), and that some clinical response can be observed (PF-05082566: stable disease). That’s a pretty good picture coming out of Phase 1. As preclinical data suggest that 4-1BB is most effective in various combination formats, it will be interesting to monitor advances in the rituximab and cetuximab co-therapy arms of these trials. Several potent combinations arising in the preclinical literature include 4-1BB with immune checkpoint inhibition (CTLA4 or the PD-1 pathway) and in combination with agonist OX40 antibody therapy.

Lets get back to the Treg cells, whose function is to suppress immune responses, primarily those of CD4+ T cells. These express 4-1BB constitutively, although it’s not clear how or if they are responding to treatment with agonist anti-4-1BB antibodies. Let’s turn to a different pathway known to have a profound effect on Tregs, the Glucocorticoid-Induced TNFR Related gene (GITR). GITR was first identified as a regulatory T-cell marker and was shown to play a critical role in breaking T cell tolerance by direct suppression of Treg activity. The preclinical evaluation of GITR produced some very striking data, including in combination settings in which anti-GITR antibodies essentially synergized with other immune checkpoint therapeutics to eliminate established tumors. Such combinations have included PD-1 blockade and CTLA4 blockade. GITR agonism is also synergistic with chemotherapy in preclinical models. Clinical development of GITR antibodies has been slow. A program initiated at TolerX and reborn at GITR, Inc., is recruiting for a phase 1 trial in advanced melanoma and other solid tumors. The antibody is TRX518 (NCT01239134). Merck is advancing a phase 1 study with the anti-GITR antibody MK-4166, although this trial (NCT02132754) is not yet recruiting patients. Review articles have mentioned an ongoing clinical program at MSKCC – this is one of the three sites enrolling patients in the TRX518 trial.

To the extent that the driving mechanism of action (MOA) of GITR stimulation is shown to be downregulation of Treg activity, this pathway should be a good candidate for combination therapy with 4-1BB, OX40 or CD27 agonists (see below) as well as with the CTLA4 and PD-1 pathway antagonists. If the MOA in human cancer patients is different or more complex than proposed, different combinations may be more or less attractive.

One last receptor – CD27.

The costimulatory molecule CD27 is constitutively expressed on most effector T cells, memory B cells, and an NK cell subset. So its expression may also overlap with those of the other receptors. CD27 appears to be important for sustained T cell effector function and also the development of T cell memory. CD27 is a marker of memory T cells, conversely, it is low or absent on Tregs. More broadly, CD27 supports germinal center formation that drives B cell maturation and the differentiation of plasma cells that produce high affinity antibodies, and is also important in driving the cytolytic activity of some NK cells.

Although there is a large preclinical literature on CD27 and its ligand CD70, there are few antibodies in clinical development. There are several historical explanations for this I think. CD70 is expressed at high levels on certain tumor types, particularly renal cell carcinoma (RCC). Much effort has gone into the development of cell-depleting antibodies targeting CD70. This expression pattern also called into question the relevance of CD27 in controlling tumor growth, as the ligand would be expected to stimulate immune responses. We now know that RCC and other solid tumors expresses high levels of PD-L1, and likely disables immune responses via this pathway. Not surprisingly then, one of the ongoing clinical efforts is a combination trial of nivolumab, the anti-PD-1 antibody from BMY with CDX-1127, an anti-CD27 antibody from Celldex (CLDX) in a collaboration announced by the 2 companies last week. In the meantime we have 2 abstracts from CLDX at ASCO in June to look forward to. In a 3×3 phase 1 dose escalation study of B cell lymphoma patients the drug was well tolerated with weekly IV dosing, and there were signs of clinical response, including a durable complete response in one patient with advanced refractory disease (abstract #3024; clinical trial NCT01460134). In the same trial, solid tumor patients were treated in a dose escalation phase and then an expansion phase (RCC and melanoma). The drug was well tolerated, there were preliminary signs of clinical response, and measureable activation of the immune system (Abstract #3027). With the potential to support memory T cell differentiation, CD27 may provide an important additional signal to drive long term tumor control. We’ll have to wait and see.

So, we have 4 receptors with overlapping activities and we have multiple antibodies in various stages of development. There will be plenty to learn about these targets and their roles in the future of combination immunotherapy. One of the most promising paths forward is the analysis of immune checkpoint and costimulatory proteins on tumor infiltrating lymphocytes (TILs). It seems very likely that the makeup of the tumor cell defense against the immune system will diverge between tumor types, and perhaps between patients with the same tumor types, or even with the same patient tumor at different times, or in different metastatic locations. Profiling TILs, and perhaps sentinel lymph nodes, for the expression patterns of lymphocytes and antigen presenting cells is likely to help guide combination therapy.

We’ll come back to that. And we’ve not forgotten those NK cells either.

stay tuned.